Next Article in Journal
Dysfunction in the Cystic Fibrosis Transmembrane Regulator in Chronic Obstructive Pulmonary Disease as a Potential Target for Personalised Medicine
Previous Article in Journal
Microbiome Modulation as a Therapeutic Approach in Chronic Skin Diseases
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nanostructures as Targeted Therapeutics for Combating Oral Bacterial Diseases

by
Shima Afrasiabi
1,†,
Nasim Chiniforush
1,†,
Hamid Reza Barikani
2,
Alireza Partoazar
3,* and
Ramin Goudarzi
4,*
1
Laser Research Center of Dentistry, Dentistry Research Institute, Tehran University of Medical Sciences, Tehran 1441987566, Iran
2
Dental Implant Research Center, Dental Research Institute, Tehran University of Medical Sciences, Tehran 1441987566, Iran
3
Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran 1441987566, Iran
4
Division of Research and Development, Pharmin USA, LLC, San Jose, CA 95128, USA
*
Authors to whom correspondence should be addressed.
Equal contribution.
Biomedicines 2021, 9(10), 1435; https://doi.org/10.3390/biomedicines9101435
Submission received: 16 September 2021 / Revised: 2 October 2021 / Accepted: 5 October 2021 / Published: 10 October 2021
(This article belongs to the Section Molecular and Translational Medicine)

Abstract

:
Pathogenic oral biofilms are now recognized as a key virulence factor in many microorganisms that cause the heavy burden of oral infectious diseases. Recently, new investigations in the nanotechnology field have propelled the development of novel biomaterials and approaches to control bacterial biofilms, either independently or in combination with other substances such as drugs, bioactive molecules, and photosensitizers used in antimicrobial photodynamic therapy (aPDT) to target different cells. Moreover, nanoparticles (NPs) showed some interesting capacity to reverse microbial dysbiosis, which is a major problem in oral biofilm formation. This review provides a perspective on oral bacterial biofilms targeted with NP-mediated treatment approaches. The first section aims to investigate the effect of NPs targeting oral bacterial biofilms. The second part of this review focuses on the application of NPs in aPDT and drug delivery systems.

1. Introduction

Biofilms have an important impact on humans in many ways as they can develop in natural, medical, and industrial environments [1]. The oral cavity is an ideal environment for microbial cell growth, survival, and stability, followed by oral biofilm formation on the tooth surface. Biofilms contain a set of bacteria that are produced in extracellular polymeric substances (EPS). Bacterial growth results in the conversion of bacterial biofilm from commensal plaque to a pathogenic form. Bacteria present in the biofilm are significantly less sensitive to antimicrobial agents than planktonic bacteria [2]. In addition, biofilm-associated cells differ in physiology and trigger different gene expression profiles to adapt to the biofilm environment [3]. For example, the genes SMU.629 and SMU.1591 encode superoxide dismutase and catabolite control protein A, which are involved in the oxidative stress tolerance and the expression of biofilm-associated genes in Streptococcus mutans, respectively. SMU.629 and SMU.1591 were impressively increased in biofilm [4,5].
The bacterial composition of dental biofilms is relatively stable and mainly consists of sites that are protected from biofilm-removing forces applied in the mouth [6]. Biofilms formed on the teeth produce diseases such as caries, chronic gingivitis, and periodontitis that have a wide range of side effects. If dental biofilms persist, they become harder and form calculus in the lower and upper gum areas, which cause an inflammatory response of the host and increase the amounts of tissue fluid flowing into gingival pockets and can lead to gum disease [7].
The inhibition of oral bacterial biofilms is challenging. The low pH milieu indicative of oral biofilms reduces the efficacy of antibiotics on oral biofilms. The acidic environment induces further EPS synthesis. The presence of EPS reduces drug access and triggers bacterial tolerance to antibiotics [8]. The mechanical removal of bacterial biofilms from dental surfaces is a proven method of treating periodontal diseases. However, mechanical procedures alone are not able to entirely kill all bacteria. Furthermore, the use of metal instruments to remove oral biofilms will result in tooth structure removal over time, and can ultimately lead to gum resorption and tooth sensitivity to physical and thermal stimulation-induced effects [9]. It has also been reported that chlorhexidine has many adverse effects ranging from mild symptoms such as headache and dry mouth to severe symptoms such as tooth staining, calculus formation, and upper respiratory tract infection and, therefore, is not suitable for daily, long-term use. Alternative anti-biofilm agents include alcoholic mouthwashes, which can irritate the oral mucosa and lead to oral cancer [10]. Additionally, natural therapeutic drugs have little efficacy due to poor drug solubility, and low penetration of the EPS matrix [8].
An alternative way for infection control has been the design and development of novel biocompatible and non-absorbable nanoparticles (NPs), which may reach high local bioactivity in a controlled release of antibacterial effects [11]. NPs have distinct advantages in that they exhibit special physical and chemical properties due to their ultra-small sizes and large surface area-to-mass ratio. This includes increased reactivity, greater solubility, biomimetic features and the ability to be functionalized with other substances such as drugs, bioactive molecules and photosensitizers. Furthermore, antimicrobial NPs can effectively infiltrate the oral biofilms, leading to the effective delivery of therapeutics and may help manage the use of antibiotics [12]. The large surface area and high charge density of NPs enable them to interact with negatively charged bacterial cells, causing enhanced antimicrobial activity, cell membrane damage, generation of reactive oxygen species (ROS), interference with cellular processes, proteins destruction, and, finally, cell death induced by DNA damage [13]. Despite several benefits, NPs have some disadvantages, such as toxicity, refraining from the physiological barrier, evading rapidly from the phagocytic cells, and mounting an immune response that should be carefully considered if they are used in living organisms [14].
Antimicrobial photodynamic therapy (aPDT), which combines a photosensitizer and a light source, has been suggested as an alternative technique for bacteria inactivation. To perform efficient aPDT, photosensitizers must be able to infiltrate bacteria [15]. NPs improve the effectiveness of the photosensitizer because they can pass through the bacteria cell walls [16]. Additionally, nanotechnology in the case of drug delivery vehicles can improve drug availability into the target tissue to deliver the maximum therapeutic effect [17]. Herein, this review investigated the antimicrobial actions of NPs against oral infectious disease with a focus on oral bacterial biofilms. Additionally, we will also elaborate on the role of NPs when applied in aPDT and drug delivery systems.

2. Methods

An electronic search through Scopus, PubMed, Web of Science and Google Scholar databases were conducted. All databases were searched from their inception to 15 September 2021. The following terms were searched in combination: nanoparticles, biofilms, dental caries, root canal therapy, periodontitis, antimicrobial photodynamic therapy, and drug delivery systems. Then, full-text articles were read for a complete assessment and determination of inclusion or exclusion. The inclusion criteria for our review were all types of articles published in English involving the keywords.

3. NPs and Their Role in Dental Caries Control

Dental caries is a multifactorial and biofilm-mediated oral disease [18]. The basic mechanism of dental caries is an ecological shift within the dental biofilm to acidogenic bacteria growth, frequently made by exposure to fermentable carbohydrates. The change in pH leads to an imbalance between de/remineralization processes, thus creating clinical decay [19]. S. mutans is the major bacterial species of dental caries [20], and it is capable of reducing environmental pH that results in the decalcification of the tooth [20]. Controlling dental caries can, therefore, be achieved through preventing the bacterial action, helping reverse tooth demineralization, and promoting the remineralization process [21]. The complete removal of carious dentin before placing a restoration often results in a pulpal exposure [22]. According to scientific evidence, every effort ideally should be made to maintain viable pulp tissue [23]. The use of nanostructures in dental caries management has also received much attention [24]. One study explored how chitosan (CS) combined with zinc oxide/zeolite (ZnO/Z) nanocomposite accelerates the biofilm reduction, metabolic activity and cariogenic properties of S. mutans [25]. The effect of ZnONPs on bacteria is through damage to the cell wall, lipids and protein, resulting in the leakage of intracellular contents to the outside of the bacteria and, ultimately, bacterial death [26].
Furthermore, recurrent caries is a major factor for restoration failure [27]. To address this problem, an adhesive containing NPs of amorphous calcium phosphate (NACP) was developed. NACP could enhance the Ca and P ions released under low pH conditions. Moreover, NACP adhesive could rapidly change the solution pH from 4.0 to above 5.5, improving the dentin remineralization. The NACP adhesive was also reported to show inhibition properties against bacteria and decrease the colony-forming units count in a biofilm of S. mutans [28].
Furthermore, metallic NPs are popular due to their antibacterial properties and biocidal activities at low concentrations. Copper NPs (CuNPs) and ZnONPs have been demonstrated to be effective against a wide range of bacteria. The addition of CuNP/ZnONP in an adhesive system provides more antimicrobial activities, without affecting their bond strength. Moreover, CuNP/ZnONP also inhibited matrix metalloprotease-2, and may improve enamel remineralization [29]. Matrix metalloprotease-2 is associated with the development of dental germ as well as the progression of dental caries in patients with necrotic pulp [30].
Another pertinent example is that of Cao and colleagues, who used a novel resin-based dental composite containing a photocurable core–shell of silver bromide (AgBr) plus cationic polymer nanocomposite (AgBr/BHPVP). The potent antibacterial effects of this resin composite against S. mutans are exerted through its biocide-releasing mechanism of the Ag+ ions. Thus, resins containing AgBr/BHPVP NPs with long-term antimicrobial effects exhibited excellent potential as an antimicrobial agent which could be used in dental products [31].

4. NPs and Their Role in Root Canal Treatment

The failure of root canal treatment is attributed to the eradication of bacteria and incomplete disinfection of the complex root canal system, which will inevitably lead to persistent apical periodontitis. Enterococcus faecalis is a bacterium often isolated from the root-filled teeth with chronic apical periodontitis [32]. E. faecalis invades the dentinal tubules, adheres to the root canal wall and forms a biofilm on dentin [32]. Endodontic diseases are primarily associated with biofilm-mediated infection [33]. Microbial biofilms inside the root canal are very resistant to conventional medicaments including systemic antibiotics and sealers. The anatomical complexities and the multi-species biofilms increase the difficulty in the complete eradication of E. faecalis microbial biofilm. In recent years, the application of NPs to disinfect root canals has gained attention due to their broad spectrum antibacterial activity [34].
A study investigated the antibacterial activity of propolis NPs (300 μg/mL) with an average size of 117.6 nm as a root canal irrigant against a root canal infected with E. faecalis biofilm. Results confirmed propolis NPs were equally effective as NaOCl (6%) and chlorhexidine (2%) in reducing the E. faecalis biofilm [35]. Interestingly, an in vitro study exhibited promising results on root canal surfaces treated with cationic antibacterial NPs such as ZnONP, CS/ZnONP, or CS-layer-ZnONP. But this effect was not considered efficient enough to elicit whether the inhibition of bacterial recolonization and biofilm formation was due to the killing of bacteria or by the direct effect of NPs on the bacteria–substrate interaction [36].
Recently, a novel antibacterial root canal sealer (dimethylaminohexadecyl methacrylate (DMAHDM)), and NACP were developed and tested on E. faecalis biofilm inhibition. There was a 3-log reduction in E. faecalis counts due to the new sealer compared to the control. Moreover, this root canal sealer demonstrated acid-neutralizing capabilities which could be useful in preventing anaerobic growth. The therapeutic root canal sealer, by releasing Ca and P ions, was able to inhibit E. faecalis biofilms. This structure, with antibacterial effects and remineralization capabilities, is a promising strategy to improve the success rate of endodontic therapy and dentin hardness [37].

5. NPs and Their Role in Periodontitis and Peri-Implantitis

Periodontitis, a chronic inflammatory condition, usually gradually destroys the tooth-supporting structures, subsequently leading to the looseness and, finally, the loss of teeth. Periodontitis is not only considered a major risk of further tooth loss, but is also reported to be associated with systemic disorders [38]. Periodontitis is one of the main risk factors that enhance the risk for peri-implantitis [39]. Peri-implantitis is an inflammatory disease affecting the tissues surrounding osseointegrated dental implants resulting in pocket formation, purulence and the loss of supporting bone, which is associated with the reduction of implant survival [40,41]. The accumulation of bacteria on the surface of implant and multi-species biofilm formation is considered the cause of dental implant failure and peri-implantitis [41].
Porphyromonas gingivalis, Prevotella intermedia and Aggregatibacter actinomycetemcomitans are the top three periodontitis and peri-implantitis-associated species in subgingival plaque. In the periodontal pockets, these bacteria can regulate the expression of many virulence factors that lead to supporting bone loss [42]. Recent studies have recently stated significant transcriptional changes when the above-mentioned bacteria were growing within a microbial multi-species biofilm [43,44,45]. Despite comprehensive mechanical and antimicrobial treatment, the long-term control of infection is impossible [46]. The use of NPs in the periodontal field is fascinating, particularly for infection control and oral biofilm management [47].
Carbon quantum dots are a new type of carbon-based nanomaterials which have unique properties such as high photostability, favorable water solubility, biocompatibility, low toxicity, and ease of preparation and modification. Liang and colleagues fabricated tinidazole carbon quantum dots using the hydrothermal method. The antimicrobial activity of tinidazole carbon quantum dots depends on its ability to impair toxicity by inhibiting the main virulence factors associated with the biofilm formation of P. gingivalis and, hence, affecting the self-assembly process of biofilm-related proteins [48].
Nanoscale surface modification has been suggested to affect bacterial adherence and biofilm formation on implants. Besinis et al. confirmed that a dual-layered silver plus nano-hydroxyapatite (HA) coating exhibited a strong inhibitory effect on the growth of Streptococcus sanguinis in the surrounding media and reduced the biofilm on the titanium-based implant surface by 97.5% [49]. S. sanguinis plays a significant role as the initial colonizer in the early adhesion stage of biofilm formation and titanium implant colonization, because it bonds directly to the implant surface and facilitates the later bacterial adhesion [50]. This novel nanocoating on titanium implants decreased infection risk, improved osseointegration, and enhanced bone healing [49].

6. NPs and Their Role in Orthodontics Infection Control

Fixed orthodontic treatments hamper the proper cleaning process, leading to biofilm buildup and white spot lesion formation. After orthodontic therapies with fixed appliances, there are significant increases in the amounts of cariogenic bacteria and biofilm formation [51]. Adding NPs to the orthodontic adhesives can reduce the frictional force between the orthodontic bracket and wire, improve the antimicrobial properties, and prevent enamel demineralization during orthodontic treatment [52].
The inhibitory effect of silver NPs in biofilm growth could predict that the silver NPs reduce biofilm metabolic activity [53]. Additionally, Xie et al. reported that the dental composite containing NACP considerably inhibited enamel demineralization and white spot lesions around orthodontic brackets [54]. Another study showed that orthodontic composites containing 1% titanium oxide NPs could significantly reduce the number of bacterial cells compared with conventional composites, without compromising the shear bond strength [55].

7. NPs and Their Role in Denture Base Material

The use of removable partial and complete dentures acts as a factor that favors bacterial accumulation in the oral cavity. The use of denture base material that possesses an antimicrobial effect may prevent future recurring infections by managing microbial colonization and halting biofilm formation [56]. Poly (methyl methacrylate) (PMMA) has been used as a principle polymer-based material for dentures. Titanium dioxide NP with antimicrobial properties against a wide range of bacteria has been recommended as filler in polymeric materials [57]. Anehosur and co-workers found that PMMA containing 3% titanium dioxide NP decreased the amount of Staphylococcus aureus, prevented quorum sensing, and eventually stopped biofilm formation on denture surface [58]. Likewise, Bacali and co-workers investigated the addition of graphene silver NP to PMMA acrylic resins in an attempt to improve denture antimicrobial characteristics and found that this combination restricted the growth of P. gingivalis and E. faecalis. In addition, the use of extra aPDT showed a greater effect on the growth inhibition of these two bacterial strains [59]. A summary of NP applications developed against oral bacterial infections are listed in Table 1.

8. Applications of NPs in aPDT

Antimicrobial photodynamic therapy (aPDT) has become an attractive alternative to traditional treatment thanks to outstanding advantages such as its harmless visible light, ease of use, and, above all, its sensitivity to ROS-mediated killing in microorganisms [81]. The employment of aPDT involved a light source under a specific wavelength in combination with photosensitizers and oxygen, which allowed an attack against microorganisms by producing ROS. The singlet photosensitizer in the ground state was extremely unstable and could instantaneously release energy, generating the triplet state. The released energy was absorbed by the tissue oxygen to form ROS, which had strong oxidative stress and cell death [82] (Figure 1).
Studies have found that aPDT exhibits an efficient bactericidal performance against oral bacteria [83,84,85,86,87]. Fumes et al. investigated aPDT’s effect on S. mutans using methylene blue (MB) as the photosensitizer and an energy density of 6 J/cm2. The results showed that aPDT can effectively decrease the viability of S. mutans biofilms [84]. Goulart et al. examined the efficacy of aPDT by using MB and erythrosine and an energy density of 0.65–2 J/cm2 to inactivate A. actinomycetemcomitans. The decrease in the A. actinomycetemcomitans biofilm in the presence of MB and erythrosine was 54% and 77%, respectively [85]. Street et al. demonstrated that the ability of aPDT, by using MB and an energy dose of 9.4 J/cm2, in eradicating the biofilm form of Fusobacterium nucleatum, P. gingivalis, and A. actinomycetemcomitans [86]. López-Jiménez et al. also demonstrated the effect of aPDT using MB or toluidine blue O (TBO) with an energy density of 271 and 106.4 J/cm2, respectively, on disrupting E. faecalis biofilms [87].
Nevertheless, the drawbacks of conventional photosensitizers, including poor solubility in aqueous media, limited in vivo stability, low extinction coefficient, and poor target selectivity, had hampered the clinical applications. To overcome these challenges, nanomaterials have been shown to be highly efficient and safe when complexed with photosensitizers. They possess important properties: controlled release of photosensitizers, enhanced water solubility, the ability to prevent aggregation even at high concentrations, excellent binding, improved ROS production, and increased bacterial cell wall penetrability [88] (Figure 2). Furthermore, nanosystems can deliver photosensitizers into cells and protect them from light, temperature, pH, and enzymatic degradation, and can decrease high-energy doses of light as well as the frequency of the administration of photosensitizers [89]. In recent years, NP-based aPDT has become a hot topic in antibacterial therapy [42].
In previous work by our group, propolis NPs in combination with PhotoActive+ and TBO as photosensitizers indicated higher antibacterial activity during aPDT against S. mutans than the individual photosensitizer. Furthermore, the expression level of genes that play an important role in S. mutans biofilm formation was also decreased to approve the high efficiency of the proposed NPs in combination with photosensitizers [90].
In another study, aPDT with MB-poly(lactic-co-glycolic) [PLGA] cationic NPs (50 µg/mL and 30 J/cm2) in chronic periodontitis patients reduced bacterial viability in both planktonic and biofilm lifestyles by approximately 60% and 48%, respectively [91]. Perhaps it can be concluded that the MB-NP was able to diffuse MB dye within biofilms [92]. A summary of NP applications in aPDT against oral bacterial infections are listed in Table 2.

9. Application of NPs in Drug Delivery Systems

The effective drug concentration reached within the biofilm is very crucial [17]. The impaired diffusion of antimicrobial agents in the biofilm mode of growth is another cause of antimicrobial therapy failure. Restricted penetration of the drug is, on the one hand, caused by cell–cell interactions within the biofilm matrix and, on the other hand, bacterial cells that exist in a dormant state and are organized into structured communities embedded within an extracellular matrix [100].
Currently, nano-scale drug delivery systems have emerged as attractive platforms. They could improve therapeutic effects and decrease the unwanted after-effects compared with conventional medicine [101]. Nanocarriers should be able to encapsulate drugs efficiently, while having the ability to increase drug transport to the deep biofilm structure [100]. One example is ciprofloxacin (CIP) in biofilms related to root canal infections. In a recent study, antibacterial and anti-biofilm activity of CIP-loaded PLGA NPs and CIP-PLGA NPs coated with CS (CIP-PLGA/CS) versus CIP solution as a control on E. faecalis was evaluated. CIP-PLGA/CS showed lower release (78.3%) than the CIP in the first hour (100%). CIP-PLGA/CS showed the highest decrease in biofilm inhibition (86.4%) which indicated that CS NPs are able to penetrate the biofilm and promote antibacterial effects. In addition, CIP-PLGA/CS, dramatically declined the count of E. faecalis strains in all concentrations [32].
The drug delivery systems in periodontal pockets had a beneficial therapeutic application for periodontitis [101]. In one study, antibacterial activity of the free minocycline and minocycline-PEGylated PLGA NPs were examined against A. actinomycetemcomitans. The results demonstrated that the encapsulation of minocycline into the PEGylated PLGA NPs increased the antibacterial efficiency against A. actinomycetemcomitans 2-fold, compared to that of the drug alone. This was possibly due to an increase in the penetration of the NPs into the microbial community and a better delivery of minocycline to the target cells [102]. Similarly, tetracycline-loaded titanium dioxide nanotube revealed excellent antibacterial and anti-adherence effects against P. gingivalis [103].
Nanocarriers can increase the beneficial effects of aPDT by increasing stability, enhancing the selectivity of treatment and inhibiting aggregation known to reduce the phototoxicity commonly seen in the treatment. The nonspecific distribution of the photosensitizer after administration represents a major obstacle to drug efficacy in aPDT. In addition, some of the photosensitizers are degraded in photolytic and hydrolytic stress conditions or metabolized after administration [104].
The results of a study showed nanocarriers with enhanced antibacterial effects could improve the efficiency of photosensitizers. The efficacy of a photosensitizer loaded with various nanocarriers including GO, GO-carnosine (Car) and GO-Car/HA were investigated against S. mutans via irradiation with an 810 nm diode laser system [105]. Graphene oxide (GO), a one atom thick molecule, easily adsorbs polymers and proteins, negatively interfering with microorganisms’ colonization, aggregation and their initial adhesion to surfaces. These interesting, recognized GO properties represent a promising approach for antimicrobial and anti-biofilm applications [106]. GO-indocyanine green (ICG), GO-Car-ICG and GO-Car/HA-ICG-mediated aPDT significantly declined the count of S. mutans. They also reduced biofilm formation by more than 50%. The expression of the gtfB gene was considerably reduced several-fold after aPDT was in the presence of the formulation. It could be concluded that the GO exhibited great potential to yield high stability and increase the inhibitory effects of ICG-mediated aPDT against S. mutans [105].
Hydrogel-based drug delivery systems have also received considerable attention [107]. NP-loaded hydrogels can not only enhance the bioavailability, efficacy and safety of the drugs, but also achieve higher mechanical strength and extend the drug in the target tissue [108,109]. Recently, Johnson et al. developed an antimicrobial-loaded nanofiber hydrogel-based drug delivery system composed of cellulose nanofibers and κ-carrageenan oligosaccharides NPs for the treatment of periodontitis. This material has strong antibacterial activity against periodontal pathogens. It has been reported that this material helps to reduce ROS generation and the cytokines production level under inflammatory situations [107].

10. Conclusions and Perspectives

The human oral cavity has been found to be an environment with bacterial diversity. In line with this, dysbiosis in the oral cavity is linked with many diseases such as dental caries, root canal infection, and periodontal disease. Overall, it can be said that the NPs are a valid therapeutic modality that can feasibly be implemented in the oral healthcare system. The potentiation of aPDT by NPs is one such candidate for effective antimicrobial therapies, as demonstrated in this review. The scope of future research lies in the development of NP technologies for the therapeutic management of oral infections and the evaluation of models to re-evaluate the removal of the biomass from the teeth, in addition to assessing the action of novel antimicrobial agents on multiple-species biofilm. The scarcity of clinical studies highlights the need for more in vivo investigations. Thus, we recommend more regular steps to be taken in order to put the nanostructure-based formulations that have been proposed on the market.

Author Contributions

S.A. contributed to study design and data gathering, wrote the manuscript and drew the figures, and reviewed and edited the final manuscript; N.C. contributed to reviewing and edited the final manuscript; H.R.B. visualization; A.P. contributed to study design and editing of the manuscript; R.G. project administration. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

All authors gave their consent for publication.

Conflicts of Interest

The authors declare that they have no competing interest.

References

  1. López, D.; Vlamakis, H.; Kolter, R. Biofilms. Cold Spring Harb. Perspect. Biol. 2010, 2, a000398. [Google Scholar] [CrossRef] [PubMed]
  2. Feuerstein, O. Light therapy: Complementary antibacterial treatment of oral biofilm. Adv. Dent. Res. 2012, 24, 103–107. [Google Scholar] [CrossRef] [PubMed]
  3. Feng, G.; Klein, M.I.; Gregoire, S.; Singh, A.P.; Vorsa, N.; Koo, H. The specific degree-of-polymerization of A-type proanthocyanidin oligomers impacts Streptococcus mutans glucan-mediated adhesion and transcriptome responses within biofilms. Biofouling 2013, 29, 629–640. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Wen, Z.T.; Baker, H.V.; Burne, R.A. Influence of BrpA on critical virulence attributes of Streptococcus mutans. J. Bacteriol. 2006, 188, 2983–2992. [Google Scholar] [CrossRef] [Green Version]
  5. Browngardt, C.M.; Wen, Z.T.; Burne, R.A. RegM is required for optimal fructosyltransferase and glucosyltransferase gene expression in Streptococcus mutans. FEMS Microbiol. Lett. 2004, 240, 75–79. [Google Scholar] [CrossRef] [Green Version]
  6. Metwalli, K.H.; Khan, S.A.; Krom, B.P.; Jabra-Rizk, M.A. Streptococcus mutans, Candida albicans, and the human mouth: A sticky situation. PLoS Pathog. 2013, 9, e1003616. [Google Scholar] [CrossRef] [Green Version]
  7. Decker, E.-M.; Klein, C.; Schwindt, D.; Von Ohle, C. Metabolic activity of Streptococcus mutans biofilms and gene expression during exposure to xylitol and sucrose. Int. J. Oral Sci. 2014, 6, 195–204. [Google Scholar] [CrossRef]
  8. Benoit, D.S.; Sims, K.R., Jr.; Fraser, D. Nanoparticles for Oral Biofilm Treatments. ACS Nano 2019, 13, 4869–4875. [Google Scholar] [CrossRef]
  9. Dosselli, R.; Millioni, R.; Puricelli, L.; Tessari, P.; Arrigoni, G.; Franchin, C.; Segallaa, A.; Teardoa, E.; Reddi, E. Molecular targets of antimicrobial photodynamic therapy identified by a proteomic approach. J. Proteom. 2012, 77, 329–343. [Google Scholar] [CrossRef] [PubMed]
  10. Sahlan, M.; Ardiellaputri, A.P.; Hermasnyah, H.; Wijanarko, A.; Darwita, R.R. Anti oral biofilm mouthwash nanoemulsion containing extract propolis and curcumin. In Proceedings of the AIP Conference Proceedings, Bante, Indonesia, 1–2 November 2018; Volume 2024, p. 020059. [Google Scholar] [CrossRef]
  11. Sanchez, M.; Toledano-Osorio, M.; Bueno, J.; Figuero, E.; Toledano, M.; Medina-Castillo, A.; Osorio, R.; Herreraa, D.; Sanz, M. Antibacterial effects of polymeric PolymP-n Active nanoparticles. An in vitro biofilm study. Dent. Mater. 2019, 35, 156–168. [Google Scholar] [CrossRef] [PubMed]
  12. Wong, J.; Zou, T.; Lee, A.H.C.; Zhang, C. The Potential Translational Applications of Nanoparticles in Endodontics. Int. J. Nanomed. 2021, 16, 2087–2106. [Google Scholar] [CrossRef]
  13. Cao, W.; Zhang, Y.; Wang, X.; Li, Q.; Xiao, Y.; Li, P.; Wang, L.; Ye, Z.; Xing, X. Novel resin-based dental material with anti-biofilm activity and improved mechanical property by incorporating hydrophilic cationic copolymer functionalized nanodiamond. J. Mater. Sci. Mater. Med. 2018, 29, 1–13. [Google Scholar] [CrossRef]
  14. Yetisgin, A.A.; Cetinel, S.; Zuvin, M.; Kosar, A.; Kutlu, O. Therapeutic nanoparticles and their targeted delivery applications. Molecules 2020, 25, 2193. [Google Scholar] [CrossRef]
  15. Soliman, N.; Sol, V.; Ouk, T.-S.; Thomas, C.M.; Gasser, G. Encapsulation of a Ru (II) polypyridyl complex into polylactide nanoparticles for antimicrobial photodynamic therapy. Pharmaceutics 2020, 12, 961. [Google Scholar] [CrossRef]
  16. Maliszewska, I.; Wanarska, E.; Thompson, A.C.; Samuel, I.D.; Matczyszyn, K. Biogenic Gold Nanoparticles Decrease Methylene Blue Photobleaching and Enhance Antimicrobial Photodynamic Therapy. Molecules 2021, 26, 623. [Google Scholar] [CrossRef]
  17. Da Costa, D.; Exbrayat-Héritier, C.; Rambaud, B.; Megy, S.; Terreux, R.; Verrier, B.; Primard, C. Surface charge modulation of rifampicin-loaded PLA nanoparticles to improve antibiotic delivery in Staphylococcus aureus biofilms. J. Nanobiotechnol. 2021, 19, 1–17. [Google Scholar] [CrossRef] [PubMed]
  18. Pitts, N.B.; Zero, D.T.; Marsh, P.D.; Ekstrand, K.; Weintraub, J.A.; Ramos-Gomez, F.; Tagami, J.; Twetman, S.; Tsakos, G.; Ismail, A. Dental caries. Nat. Rev. Dis. Primers 2017, 3, 1–16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Schwendicke, F.; Dörfer, C.; Paris, S. Incomplete caries removal: A systematic review and meta-analysis. J. Dent. Res. 2013, 92, 306–314. [Google Scholar] [CrossRef] [PubMed]
  20. Feldman, M.; Sionov, R.; Smoum, R.; Mechoulam, R.; Ginsburg, I.; Steinberg, D. Comparative Evaluation of Combinatory Interaction between Endocannabinoid System Compounds and Poly-L-lysine against Streptococcus mutans Growth and Biofilm Formation. Biomed. Res. Int. 2020, 2020, 7258380. [Google Scholar] [CrossRef]
  21. Neel, E.A.A.; Bozec, L.; Perez, R.A.; Kim, H.-W.; Knowles, J.C. Nanotechnology in dentistry: Prevention, diagnosis, and therapy. Int. J. Nanomed. 2015, 10, 371–394. [Google Scholar] [CrossRef] [Green Version]
  22. Croft, K.; Kervanto-Seppälä, S.; Stangvaltaite, L.; Kerosuo, E. Management of deep carious lesions and pulps exposed during carious tissue removal in adults: A questionnaire study among dentists in Finland. Clin. Oral Investig. 2019, 23, 1271–1280. [Google Scholar] [CrossRef] [Green Version]
  23. Bourguignon, C.; Cohenca, N.; Lauridsen, E.; Flores, M.T.; O’Connell, A.C.; Day, P.F.; Tsilingaridis, G.; Abbott, P.V.; Fouad, A.F.; Hicks, L.; et al. International Association of Dental Traumatology guidelines for the management of traumatic dental injuries: 1. Fractures and luxations. Dent. Traumatol. 2020, 36, 314–330. [Google Scholar] [CrossRef]
  24. Hannig, M.; Hannig, C. Nanotechnology and its role in caries therapy. Adv. Dent. Res. 2012, 24, 53–57. [Google Scholar] [CrossRef]
  25. Afrasiabi, S.; Bahador, A.; Partoazar, A. Combinatorial therapy of chitosan hydrogel-based zinc oxide nanocomposite attenuates the virulence of Streptococcus mutans. BMC Microbiol. 2021, 21, 1–8. [Google Scholar] [CrossRef] [PubMed]
  26. Partoazar, A.; Bideskan, F.R.; Partoazar, M.; Talaei, N.; Dallal, M.M.S. Inhibition of Biofilm Formation of Staphylococcus aureus Strains Through ZnO/Zeolite Nanocomposite and Its Cytotoxicity Evaluation. BioNanoScience 2020, 10, 714–720. [Google Scholar] [CrossRef]
  27. Melo, M.A.S.; Cheng, L.; Weir, M.D.; Hsia, R.C.; Rodrigues, L.K.; Xu, H.H. Novel dental adhesive containing antibacterial agents and calcium phosphate nanoparticles. J. Biomed. Mater. Res. B Appl. Biomater. 2013, 101, 620–629. [Google Scholar] [CrossRef] [Green Version]
  28. Tao, S.; He, L.; Xu, H.H.; Weir, M.D.; Fan, M.; Yu, Z.; Zhang, M.; Zhou, X.; Liang, K.; Li, J. Dentin remineralization via adhesive containing amorphous calcium phosphate nanoparticles in a biofilm-challenged environment. J. Dent. 2019, 89, 103193. [Google Scholar] [CrossRef]
  29. Gutiérrez, M.F.; Bermudez, J.; Dávila-Sánchez, A.; Alegría-Acevedo, L.F.; Méndez-Bauer, L.; Hernández, M.; Astorga, J.; Reis, A.; Loguercio, A.D.; Farago, P.V.; et al. Zinc oxide and copper nanoparticles addition in universal adhesive systems improve interface stability on caries-affected dentin. J. Mech. Behav. Biomed. Mater. 2019, 100, 103366. [Google Scholar] [CrossRef]
  30. Pereira Prado, V.; Asquino, N.; Apellaniz, D.; Bueno Rossy, L.; Tapia, G.; Bologna Molina, R. Metalloproteinases (MMPs) of the extracellular matrix in dentistry. Odontoestomatologia 2016, 18, 19–28. [Google Scholar]
  31. Cao, W.; Zhang, Y.; Wang, X.; Chen, Y.; Li, Q.; Xing, X.; Xiao, Y.; Peng, X.; Ye, Z. Development of a novel resin-based dental material with dual biocidal modes and sustained release of Ag+ ions based on photocurable core-shell AgBr/cationic polymer nanocomposites. J. Mater. Sci. Mater. Med. 2017, 28, 103. [Google Scholar] [CrossRef] [PubMed]
  32. Arafa, M.G.; Mousa, H.A.; Afifi, N.N. Preparation of PLGA-chitosan based nanocarriers for enhancing antibacterial effect of ciprofloxacin in root canal infection. Drug Deliv. 2020, 27, 26–39. [Google Scholar] [CrossRef] [Green Version]
  33. Elshinawy, M.I.; Al-Madboly, L.A.; Ghoneim, W.M.; El-Deeb, N.M. Synergistic effect of newly introduced root canal medicaments; ozonated olive oil and chitosan nanoparticles, against persistent endodontic pathogens. Front Microbiol. 2018, 9, 1371. [Google Scholar] [CrossRef] [Green Version]
  34. Neelakantan, P.; Romero, M.; Vera, J.; Daood, U.; Khan, A.U.; Yan, A.; Cheung, G.S.P. Biofilms in endodontics—current status and future directions. Int. J. Mol. Sci. 2017, 18, 1748. [Google Scholar] [CrossRef]
  35. Parolia, A.; Kumar, H.; Ramamurthy, S.; Madheswaran, T.; Davamani, F.; Pichika, M.R.; Mak, K.K.; Fawzy, A.S.; Daood, U.; Pau, A.; et al. Effect of Propolis Nanoparticles against Enterococcus faecalis Biofilm in the Root Canal. Molecules 2021, 26, 715. [Google Scholar] [CrossRef] [PubMed]
  36. Kishen, A.; Shi, Z.; Shrestha, A.; Neoh, K.G. An investigation on the antibacterial and antibiofilm efficacy of cationic nanoparticulates for root canal disinfection. J. Endod. 2008, 34, 1515–1520. [Google Scholar] [CrossRef]
  37. Baras, B.H.; Wang, S.; Melo, M.A.S.; Tay, F.; Fouad, A.F.; Arola, D.D.; Weir, M.D.; Xu, H.H. Novel bioactive root canal sealer with antibiofilm and remineralization properties. J. Dent. 2019, 83, 67–76. [Google Scholar] [CrossRef] [PubMed]
  38. Ni, C.; Zhou, J.; Kong, N.; Bian, T.; Zhang, Y.; Huang, X.; Xiao, Y.; Yang, W.; Yan, F. Gold nanoparticles modulate the crosstalk between macrophages and periodontal ligament cells for periodontitis treatment. Biomaterials 2019, 206, 115–132. [Google Scholar] [CrossRef]
  39. Stacchi, C.; Berton, F.; Perinetti, G.; Frassetto, A.; Lombardi, T.; Khoury, A.; Andolsek, F.; Di Lenarda, R. Risk factors for peri-implantitis: Effect of history of periodontal disease and smoking habits. A systematic review and meta-analysis. J. Oral Maxillofac. Res. 2016, 7, e3. [Google Scholar] [CrossRef]
  40. Smeets, R.; Henningsen, A.; Jung, O.; Heiland, M.; Hammächer, C.; Stein, J.M. Definition, etiology, prevention and treatment of peri-implantitis–a review. Head Face Med. 2014, 10, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Liao, J.; Anchun, M.; Zhu, Z.; Quan, Y. Antibacterial titanium plate deposited by silver nanoparticles exhibits cell compatibility. Int. J. Nanomed. 2010, 5, 337–342. [Google Scholar] [PubMed] [Green Version]
  42. Chi, M.; Qi, M.; Wang, P.; Weir, M.D.; Melo, M.A.; Sun, X.; Dong, B.; Li, C.; Wu, J.; Wang, L.; et al. Novel bioactive and therapeutic dental polymeric materials to inhibit periodontal pathogens and biofilms. Int. J. Mol. Sci. 2019, 20, 278. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Romero-Lastra, P.; Sánchez, M.C.; Llama-Palacios, A.; Figuero, E.; Herrera, D.; Sanz, M. Gene expression of Porphyromonas gingivalis ATCC 33277 when growing in an in vitro multispecies biofilm. PLoS ONE 2019, 14, e0221234. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yamanaka, T.; Furukawa, T.; Matsumoto-Mashimo, C.; Yamane, K.; Sugimori, C.; Nambu, T.; Mori, N.; Nishikawa, H.; Walker, C.B.; Leung, K.-P.; et al. Gene expression profile and pathogenicity of biofilm-forming Prevotella intermedia strain 17. BMC Microbiol. 2009, 9, 1–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Syahiran, S.; Taib, W.R.W.; Jaffar, N. Aggregatibacter actinomycetemcomitans: The virulence factors and relation to persistence biofilm formation. Biomedicine 2020, 40, 429–435. [Google Scholar] [CrossRef]
  46. Buchmann, R.; Müller, R.F.; Heinecke, A.; Lange, D.E. Actinobacillus actinomycetemcomitans in destructive periodontal disease. Three-year follow-up results. J. Periodontol. 2000, 71, 444–453. [Google Scholar] [CrossRef]
  47. Moraes, G.; Zambom, C.; Siqueira, W.L. Nanoparticles in Dentistry: A Comprehensive Review. Pharmaceuticals 2021, 14, 752. [Google Scholar] [CrossRef]
  48. Liang, G.; Shi, H.; Qi, Y.; Li, J.; Jing, A.; Liu, Q.; Feng, W.; Li, G.; Gao, S. Specific Anti-biofilm Activity of Carbon Quantum Dots by Destroying, P. gingivalis Biofilm Related Genes. Int. J. Nanomed. 2020, 15, 5473–5489. [Google Scholar] [CrossRef] [PubMed]
  49. Besinis, A.; Hadi, S.D.; Le, H.; Tredwin, C.; Handy, R. Antibacterial activity and biofilm inhibition by surface modified titanium alloy medical implants following application of silver, titanium dioxide and hydroxyapatite nanocoatings. Nanotoxicology 2017, 11, 327–338. [Google Scholar] [CrossRef] [Green Version]
  50. Aguayo, S.; Donos, N.; Spratt, D.; Bozec, L. Probing the nanoadhesion of Streptococcus sanguinis to titanium implant surfaces by atomic force microscopy. Int. J. Nanomed. 2016, 11, 1443–1450. [Google Scholar]
  51. Fernandez, C.C.; Sokolonski, A.R.; Fonseca, M.S.; Stanisic, D.; Araújo, D.B.; Azevedo, V.; Portela, R.D.; Tasic, L. Applications of Silver Nanoparticles in Dentistry: Advances and Technological Innovation. Int. J. Mol. Sci. 2021, 22, 2485. [Google Scholar] [CrossRef]
  52. Zakrzewski, W.; Dobrzynski, M.; Dobrzynski, W.; Zawadzka-Knefel, A.; Janecki, M.; Kurek, K.; Lubojanski, A.; Szymonowicz, M.; Rybak, Z.; Wiglusz, R.J. Nanomaterials Application in Orthodontics. Nanomaterials 2021, 11, 337. [Google Scholar] [CrossRef]
  53. Zhang, N.; Melo, M.A.S.; Antonucci, J.M.; Lin, N.J.; Lin-Gibson, S.; Bai, Y.; Xu, H.H.K. Novel dental cement to combat biofilms and reduce acids for orthodontic applications to avoid enamel demineralization. Materials 2016, 9, 413. [Google Scholar] [CrossRef] [Green Version]
  54. Xie, X.-J.; Xing, D.; Wang, L.; Zhou, H.; Weir, M.D.; Bai, Y.-X.; Xu, H.H. Novel rechargeable calcium phosphate nanoparticle-containing orthodontic cement. Int. J. Oral Sci. 2017, 9, 24–32. [Google Scholar] [CrossRef] [PubMed]
  55. Poosti, M.; Ramazanzadeh, B.; Zebarjad, M.; Javadzadeh, P.; Naderinasab, M.; Shakeri, M.T. Shear bond strength and antibacterial effects of orthodontic composite containing TiO2 nanoparticles. Eur. J. Orthod. 2013, 35, 676–679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ramburrun, P.; Pringle, N.A.; Dube, A.; Adam, R.Z.; D’Souza, S.; Aucamp, M. Recent Advances in the Development of Antimicrobial and Antifouling Biocompatible Materials for Dental Applications. Materials 2021, 14, 3167. [Google Scholar] [CrossRef] [PubMed]
  57. Gad, M.M.; Abualsaud, R. Behavior of PMMA denture base materials containing titanium dioxide nanoparticles: A literature review. Int. J. Biomater. 2019, 2019, 6190610. [Google Scholar] [CrossRef] [PubMed]
  58. Anehosur, G.V.; Kulkarni, R.; Naik, M.; Nadiger, R. Synthesis and determination of antimicrobial activity of visible light activated TiO2 nanoparticles with polymethyl methacrylate denture base resin against Staphylococcus aureus. J. Gerontol. Geriat. Res. 2012, 1, 1–8. [Google Scholar]
  59. Bacali, C.; Carpa, R.; Buduru, S.; Moldovan, M.L.; Baldea, I.; Constantin, A.; Moldovan, M.; Prodan, D.; Dascalu, M.L.; Lucaciu, O.; et al. Association of Graphene Silver Polymethyl Methacrylate (PMMA) with Photodynamic Therapy for Inactivation of Halitosis Responsible Bacteria in Denture Wearers. Nanomaterials 2021, 11, 1643. [Google Scholar] [CrossRef]
  60. Bhatt, L.; Chen, L.; Guo, J.; Klie, R.F.; Shi, J.; Pesavento, R.P. Hydrolyzed Ce (IV) salts limit sucrose-dependent biofilm formation by Streptococcus mutans. J. Inorg. Biochem. 2020, 206, 110997. [Google Scholar] [CrossRef]
  61. Noori, A.J.; Kareem, F.A. The effect of magnesium oxide nanoparticles on the antibacterial and antibiofilm properties of glass-ionomer cement. Heliyon 2019, 5, e02568. [Google Scholar] [CrossRef] [Green Version]
  62. Shvero, D.K.; Davidi, M.P.; Weiss, E.I.; Srerer, N.; Beyth, N. Antibacterial effect of polyethyleneimine nanoparticles incorporated in provisional cements against Streptococcus mutans. J. Biomed. Mater. Res. B Appl. Biomater. 2010, 94, 367–371. [Google Scholar] [CrossRef]
  63. Chen, C.; Weir, M.D.; Cheng, L.; Lin, N.J.; Lin-Gibson, S.; Chow, L.C.; Zhou, X.; Xu, H.H. Antibacterial activity and ion release of bonding agent containing amorphous calcium phosphate nanoparticles. Dent Mater. 2014, 30, 891–901. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kasraei, S.; Sami, L.; Hendi, S.; AliKhani, M.-Y.; Rezaei-Soufi, L.; Khamverdi, Z. Antibacterial properties of composite resins incorporating silver and zinc oxide nanoparticles on Streptococcus mutans and Lactobacillus. Restor. Dent. Endod. 2014, 39, 109–114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Bregnocchi, A.; Zanni, E.; Uccelletti, D.; Marra, F.; Cavallini, D.; De Angelis, F.; De Bellis, G.; Bossù, M.; Ierardo, G.; Polimeni, A.; et al. Graphene-based dental adhesive with anti-biofilm activity. J. Nanobiotechnol. 2017, 15, 1–13. [Google Scholar] [CrossRef] [PubMed]
  66. Akram, Z.; Aati, S.; Ngo, H.; Fawzy, A. pH-dependent delivery of chlorhexidine from PGA grafted mesoporous silica nanoparticles at resin-dentin interface. J. Nanobiotechnol. 2021, 19, 1–16. [Google Scholar] [CrossRef]
  67. Partoazar, A.; Talaei, N.; Bahador, A.; Pourhajibagher, M.; Dehpour, S.; Sadati, M.; Bakhtiarian, A. Antibiofilm activity of natural zeolite supported NanoZnO: Inhibition of Esp gene expression of Enterococcus faecalis. Nanomedicine 2019, 14, 675–687. [Google Scholar] [CrossRef]
  68. Liu, T.; Aman, A.; Ainiwaer, M.; Ding, L.; Zhang, F.; Hu, Q.; Song, Y.; Ni, Y.; Tang, X. Evaluation of the anti-biofilm effect of poloxamer-based thermoreversible gel of silver nanoparticles as a potential medication for root canal therapy. Sci. Rep. 2021, 11, 1–16. [Google Scholar]
  69. Azad, A.; Rostamifar, S.; Modaresi, F.; Bazrafkan, A.; Rezaie, Z. Assessment of the Antibacterial Effects of Bismuth Nanoparticles against Enterococcus faecalis. Biomed Res. Int. 2020, 2020, 5465439. [Google Scholar] [CrossRef]
  70. Louwakul, P.; Saelo, A.; Khemaleelakul, S. Efficacy of calcium oxide and calcium hydroxide nanoparticles on the elimination of Enterococcus faecalis in human root dentin. Clin. Oral Investig. 2017, 21, 865–871. [Google Scholar] [CrossRef]
  71. Monzavi, A.; Eshraghi, S.; Hashemian, R.; Momen-Heravi, F. In vitro and ex vivo antimicrobial efficacy of nano-MgO in the elimination of endodontic pathogens. Clin. Oral Investig. 2015, 19, 349–356. [Google Scholar] [CrossRef]
  72. Del Carpio-Perochena, A.; Bramante, C.M.; Duarte, M.A.H.; de Moura, M.R.; Aouada, F.A.; Kishen, A. Chelating and antibacterial properties of chitosan nanoparticles on dentin. Restor. Dent. Endod. 2015, 40, 195–201. [Google Scholar] [CrossRef]
  73. Leng, D.; Li, Y.; Zhu, J.; Liang, R.; Zhang, C.; Zhou, Y.; Li, M.; Wang, Y.; Rong, D.; Wu, D.; et al. The Antibiofilm activity and mechanism of nanosilver-and nanozinc-incorporated mesoporous calcium-silicate nanoparticles. Int. J. Nanomed. 2020, 15, 3921–3936. [Google Scholar] [CrossRef] [PubMed]
  74. Bukhari, S.; Kim, D.; Liu, Y.; Karabucak, B.; Koo, H. Novel endodontic disinfection approach using catalytic nanoparticles. J. Endod. 2018, 44, 806–812. [Google Scholar] [CrossRef] [PubMed]
  75. Ong, T.H.; Chitra, E.; Ramamurthy, S.; Siddalingam, R.P.; Yuen, K.H.; Ambu, S.P.; Davamani, F. Chitosan-propolis nanoparticle formulation demonstrates anti-bacterial activity against Enterococcus faecalis biofilms. PLoS ONE 2017, 12, e0174888. [Google Scholar]
  76. Miglani, S.; Tani-Ishii, N. Biosynthesized selenium nanoparticles: Characterization, antimicrobial, and antibiofilm activity against Enterococcus faecalis. PeerJ 2021, 9, e11653. [Google Scholar] [CrossRef] [PubMed]
  77. Wang, J.; Zhan, L.; Zhang, X.; Wu, R.; Liao, L.; Wei, J. Silver Nanoparticles Coated Poly (L-Lactide) Electrospun Membrane for Implant Associated Infections Prevention. Front Pharmacol. 2020, 11, 431. [Google Scholar] [CrossRef] [Green Version]
  78. Lee, D.; Lee, S.J.; Moon, J.-H.; Kim, J.H.; Heo, D.N.; Bang, J.B.; Lim, H.-N.; Kwon, I.K. Preparation of antibacterial chitosan membranes containing silver nanoparticles for dental barrier membrane applications. J. Ind. Eng. Chem. 2018, 66, 196–202. [Google Scholar] [CrossRef]
  79. Massa, M.A.; Covarrubias, C.; Bittner, M.; Fuentevilla, I.A.; Capetillo, P.; Von Marttens, A.; Carvajal, J.C. Synthesis of new antibacterial composite coating for titanium based on highly ordered nanoporous silica and silver nanoparticles. Mater. Sci. Eng. C Mater. Biol. Appl. 2014, 45, 146–153. [Google Scholar] [CrossRef]
  80. Vega-Jiménez, A.; Almaguer-Flores, A.; Flores-Castañeda, M.; Camps, E.; Uribe-Ramírez, M.; Aztatzi-Aguilar, O.; De Vizcaya-Ruiz, A. Bismuth subsalicylate nanoparticles with anaerobic antibacterial activity for dental applications. Nanotechnology 2017, 28, 435101. [Google Scholar] [CrossRef] [Green Version]
  81. Klausen, M.; Ucuncu, M.; Bradley, M. Design of photosensitizing agents for targeted antimicrobial photodynamic therapy. Molecules 2020, 25, 5239. [Google Scholar] [CrossRef]
  82. Polat, E.; Kang, K. Natural Photosensitizers in Antimicrobial Photodynamic Therapy. Biomedicines 2021, 9, 584. [Google Scholar] [CrossRef] [PubMed]
  83. Zhang, T.; Ying, D.; Qi, M.; Li, X.; Fu, L.; Sun, X.; Wang, L.; Zhou, Y. Anti-biofilm property of bioactive upconversion nanocomposites containing chlorin e6 against periodontal pathogens. Molecules 2019, 24, 2692. [Google Scholar] [CrossRef] [Green Version]
  84. Esteban Florez, F.L.; Mendonca de Oliveira, M.R.; de Oliveira Junior, O.B.; Hiers, R.D.; Khajotia, S.S.; Pretel, H. Bioluminescence analysis of antibacterial photodynamic therapy using methylene blue mediated by low-intensity level laser against cariogenic biofilms. Photomed. Laser Surg. 2018, 36, 258–265. [Google Scholar] [CrossRef]
  85. De Carvalho Goulart, R.; Thedei, G., Jr.; Souza, S.L.; Tedesco, A.C.; Ciancaglini, P. Comparative study of methylene blue and erythrosine dyes employed in photodynamic therapy for inactivation of planktonic and biofilm-cultivated Aggregatibacter actinomycetemcomitans. Photomed. Laser Surg. 2010, 28 (Suppl. S1), S85–S90. [Google Scholar] [CrossRef] [PubMed]
  86. Street, C.N.; Pedigo, L.A.; Loebel, N.G. Energy dose parameters affect antimicrobial photodynamic therapy–mediated eradication of periopathogenic biofilm and planktonic cultures. Photomed. Laser Surg. 2010, 28 (Suppl S1), S61–S66. [Google Scholar] [CrossRef]
  87. López-Jiménez, L.; Fusté, E.; Martínez-Garriga, B.; Arnabat-Domínguez, J.; Vinuesa, T.; Viñas, M. Effects of photodynamic therapy on Enterococcus faecalis biofilms. Lasers Med. Sci. 2015, 30, 1519–1526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Qi, M.; Chi, M.; Sun, X.; Xie, X.; Weir, M.D.; Oates, T.W.; Zhou, Y.; Wang, L.; Bai, Y.; Xu, H.H. Novel nanomaterial-based antibacterial photodynamic therapies to combat oral bacterial biofilms and infectious diseases. Int. J. Nanomed. 2019, 14, 6937–6956. [Google Scholar] [CrossRef] [Green Version]
  89. Silvestre, A.L.P.; Di Filippo, L.D.; Besegato, J.F.; de Annunzio, S.R.; de Camargo, B.A.F.; de Melo, P.B.G.; Rastelli, A.N.d.S.; Fontana, C.R.; Chorilli, M. Current applications of drug delivery nanosystems associated with antimicrobial photodynamic therapy for oral infections. Int. J. Pharm. 2020, 592, 120078. [Google Scholar] [CrossRef]
  90. Afrasiabi, S.; Pourhajibagher, M.; Chiniforush, N.; Bahador, A. Propolis nanoparticle enhances the potency of antimicrobial photodynamic therapy against Streptococcus mutans in a synergistic manner. Sci. Rep. 2020, 10, 1–16. [Google Scholar] [CrossRef]
  91. Klepac-Ceraj, V.; Patel, N.; Song, X.; Holewa, C.; Patel, C.; Kent, R.; Amiji, M.M.; Soukos, N.S. Photodynamic effects of methylene blue-loaded polymeric nanoparticles on dental plaque bacteria. Lasers Surg. Med. 2011, 43, 600–606. [Google Scholar] [CrossRef] [Green Version]
  92. De Freitas, L.M.; Calixto, G.M.F.; Chorilli, M.; Giusti, J.S.M.; Bagnato, V.S.; Soukos, N.S.; Amiji, M.M.; Fontana, C.R. Polymeric nanoparticle-based photodynamic therapy for chronic periodontitis in vivo. Int. J. Mol. Sci. 2016, 17, 769. [Google Scholar] [CrossRef]
  93. Sasaki, Y.; Hayashi, J.I.; Fujimura, T.; Iwamura, Y.; Yamamoto, G.; Nishida, E.; Ohno, T.; Okada, K.; Yamamoto, H.; Kikuchi, T.; et al. New irradiation method with indocyanine green-loaded nanospheres for inactivating periodontal pathogens. Int. J. Mol. Sci. 2017, 18, 154. [Google Scholar] [CrossRef]
  94. Shrestha, A.; Kishen, A. Antibiofilm efficacy of photosensitizer-functionalized bioactive nanoparticles on multispecies biofilm. J. Endod. 2014, 40, 1604–1610. [Google Scholar] [CrossRef]
  95. Sun, X.; Wang, L.; Lynch, C.D.; Sun, X.; Li, X.; Qi, M.; Ma, C.; Li, C.; Dong, B.; Zhou, Y.; et al. Nanoparticles having amphiphilic silane containing Chlorin e6 with strong anti-biofilm activity against periodontitis-related pathogens. J. Dent. 2019, 81, 70–84. [Google Scholar] [CrossRef]
  96. Okamoto, I.; Miyaji, H.; Miyata, S.; Shitomi, K.; Sugaya, T.; Ushijima, N.; Akasaka, T.; Enya, S.; Saita, S.; Kawasaki, H. Antibacterial and Antibiofilm Photodynamic Activities of Lysozyme-Au Nanoclusters/Rose Bengal Conjugates. ACS Omega 2021, 6, 9279–9290. [Google Scholar] [CrossRef]
  97. Misba, L.; Kulshrestha, S.; Khan, A.U. Antibiofilm action of a toluidine blue O-silver nanoparticle conjugate on Streptococcus mutans: A mechanism of type I photodynamic therapy. Biofouling 2016, 32, 313–328. [Google Scholar] [CrossRef] [PubMed]
  98. Peng, P.-C.; Hsieh, C.-M.; Chen, C.-P.; Tsai, T.; Chen, C.-T. Assessment of photodynamic inactivation against periodontal bacteria mediated by a chitosan hydrogel in a 3D gingival model. Int. J. Mol. Sci. 2016, 17, 1821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Ossmann, A.; Kranz, S.; Andre, G.; Völpel, A.; Albrecht, V.; Fahr, A.; Sigusch, B.W. Photodynamic killing of Enterococcus faecalis in dentinal tubules using mTHPC incorporated in liposomes and invasomes. Clin. Oral Investig. 2015, 19, 373–384. [Google Scholar] [CrossRef] [PubMed]
  100. Teirlinck, E.; Barras, A.; Liu, J.; Fraire, J.C.; Lajunen, T.; Xiong, R.; Forier, K.; Li, C.; Urtti, A.; Boukherroub, R.; et al. Exploring light-sensitive nanocarriers for simultaneous triggered antibiotic release and disruption of biofilms upon generation of laser-induced vapor nanobubbles. Pharmaceutics 2019, 11, 201. [Google Scholar] [CrossRef] [Green Version]
  101. Liang, J.; Peng, X.; Zhou, X.; Zou, J.; Cheng, L. Emerging applications of drug delivery systems in oral infectious diseases prevention and treatment. Molecules 2020, 25, 516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Kashi, T.S.J.; Eskandarion, S.; Esfandyari-Manesh, M.; Marashi, S.M.A.; Samadi, N.; Fatemi, S.M.; Atyabi, F.; Eshraghi, S.; Dinarvand, R. Improved drug loading and antibacterial activity of minocycline-loaded PLGA nanoparticles prepared by solid/oil/water ion pairing method. Int. J. Nanomed. 2012, 7, 221–234. [Google Scholar]
  103. Sun, L.; Xu, J.; Sun, Z.; Zheng, F.; Liu, C.; Wang, C.; Hu, X.; Xia, L.; Liu, Z.; Xia, R. Decreased Porphyromonas gingivalis adhesion and improved biocompatibility on tetracycline-loaded TiO2 nanotubes: An in vitro study. Int. J. Nanomed. 2018, 13, 6769–6777. [Google Scholar] [CrossRef] [Green Version]
  104. Hegge, A.B.; Bruzell, E.; Kristensen, S.; Tønnesen, H. Photoinactivation of Staphylococcus epidermidis biofilms and suspensions by the hydrophobic photosensitizer curcumin–effect of selected nanocarrier: Studies on curcumin and curcuminoides XLVII. Eur. J. Pharm. Sci. 2012, 47, 65–74. [Google Scholar] [CrossRef]
  105. Gholibegloo, E.; Karbasi, A.; Pourhajibagher, M.; Chiniforush, N.; Ramazani, A.; Akbari, T.; Bahador, A.; Khoobi, M. Carnosine-graphene oxide conjugates decorated with hydroxyapatite as promising nanocarrier for ICG loading with enhanced antibacterial effects in photodynamic therapy against Streptococcus mutans. J. Photochem. Photobiol. B 2018, 181, 14–22. [Google Scholar] [CrossRef]
  106. Di Giulio, M.; Di Lodovico, S.; Fontana, A.; Traini, T.; Di Campli, E.; Pilato, S.; D’Ercole, S.; Cellini, L. Graphene Oxide affects Staphylococcus aureus and Pseudomonas aeruginosa dual species biofilm in Lubbock Chronic Wound Biofilm model. Sci Rep. 2020, 10, 1–9. [Google Scholar] [CrossRef] [PubMed]
  107. Johnson, A.; Kong, F.; Miao, S.; Lin, H.-T.V.; Thomas, S.; Huang, Y.-C.; Kong, Z.L. Therapeutic effects of antibiotics loaded cellulose nanofiber and κ-carrageenan oligosaccharide composite hydrogels for periodontitis treatment. Sci Rep. 2020, 10, 1–23. [Google Scholar] [CrossRef] [PubMed]
  108. Sabbagh, F.; Kiarostami, K.; Mahmoudi Khatir, N.; Rezania, S.; Muhamad, I.I. Green synthesis of Mg0. 99 Zn0. 01O nanoparticles for the fabrication of κ-Carrageenan/NaCMC hydrogel in order to deliver catechin. Polymers 2020, 12, 861. [Google Scholar] [CrossRef] [Green Version]
  109. Sabbagh, F.; Kiarostami, K.; Khatir, N.M.; Rezania, S.; Muhamad, I.I.; Hosseini, F. Effect of zinc content on structural, functional, morphological, and thermal properties of kappa-carrageenan/NaCMC nanocomposites. Polym. Test. 2021, 93, 106922. [Google Scholar] [CrossRef]
Figure 1. Mechanism of action of antimicrobial photodynamic therapy. The ground state (S0) photosensitizer absorbs a photon to form the first excited singlet state (1PS) that can undergo intersystem crossing to form the first excited triplet state (3PS). The long−lived triplet state can interact with molecular oxygen in two ways: type I electron transfer to form superoxide and the hydroxyl radical, or type II energy transfer to form 1O2. The generated ROS can damage a broad spectrum of microorganisms.
Figure 1. Mechanism of action of antimicrobial photodynamic therapy. The ground state (S0) photosensitizer absorbs a photon to form the first excited singlet state (1PS) that can undergo intersystem crossing to form the first excited triplet state (3PS). The long−lived triplet state can interact with molecular oxygen in two ways: type I electron transfer to form superoxide and the hydroxyl radical, or type II energy transfer to form 1O2. The generated ROS can damage a broad spectrum of microorganisms.
Biomedicines 09 01435 g001
Figure 2. Schematic presentation of antimicrobial photodynamic therapy with nanoparticles against oral biofilm. Nanoparticles can accumulate on the membrane surface leading to local deformation and, ultimately, damaging membrane integrity. Photosensitizer-loaded nanoparticle-guided synergistic antimicrobial photodynamic therapy. After entering into the bacteria in the target site, ROS can be effectively produced under the irradiation as shown in the figure. This mechanism can improve the antimicrobial photodynamic therapy function.
Figure 2. Schematic presentation of antimicrobial photodynamic therapy with nanoparticles against oral biofilm. Nanoparticles can accumulate on the membrane surface leading to local deformation and, ultimately, damaging membrane integrity. Photosensitizer-loaded nanoparticle-guided synergistic antimicrobial photodynamic therapy. After entering into the bacteria in the target site, ROS can be effectively produced under the irradiation as shown in the figure. This mechanism can improve the antimicrobial photodynamic therapy function.
Biomedicines 09 01435 g002
Table 1. A summary of several NPs developed for combating oral bacterial infections.
Table 1. A summary of several NPs developed for combating oral bacterial infections.
NanoparticlesSize (nm)MicroorganismNanoparticle Therapeutic EffectRef.
Cerium oxide-S. mutans- Reduces adherent bacteria[60]
Magnesium oxide~20S. mutans
S. sobrinus
- Antibacterial and anti-biofilm activity[61]
Polyethyleneimine-S. mutans- Prevents caries and inflammation[62]
NACP~116Total streptococci- Increases the Ca and P ion release at cariogenic pH
- Inhibits biofilms and remineralizes tooth lesions
- Reduces metabolic activity and lactic acid of biofilm
[63]
ZnO and silver~50 and 20S. mutans
Lactobacillus
- Inhibits bacterial growth[64]
Graphene-S. mutans- Inhibits S. mutans adhesion and growth
- Anti-biofilm effects
- Application as filler in dental adhesives
[65]
MSN-PLGA~107S. mutans- Anti-biofilm effects
- Application as adhesive and restorative dentistry
[66]
ZnO/Z~30E. faecalis- Anti-biofilm effects
- Decreases esp gene expression
[67]
AgNPs-PL~21.62E. faecalis- Increases their permeability
- Destabilizes bacterial membranes
- Anti-biofilm activity
[68]
Bismuth NPs~40E. faecalis- Antibacterial activity[69]
CO and CH-E. faecalis- Antibacterial activity[70]
MgO~70–150E. faecalis- Antibacterial activity[71]
Chitosan~85–221E. faecalis- Anti-biofilm activity
- Inhibition of bacterial recolonization
[72]
Ag/Zn-MCSNs~200–250E. faecalis- Anti-biofilm activity
- Destroys cell membranes
[73]
Iron oxide-E. faecalis- Improves catalytic activity
- Enhances antibacterial activity on root canal surfaces
[74]
Chitosan-propolis~247–512E. faecalis- Disrupts the biofilm structure
- Alters the expression of biofilm-associated genes
[75]
Selenium~40–150E. faecalis- Anti-biofilm activity[76]
PLLA@Ag-S. aureus- Antibacterial activity
- Good biocompatibility
- Implant-associated infections prevention
[77]
AgCSP~300P. gingivalis- Antibacterial activity
- Good biocompatibility
- Tissue regeneration
[78]
AgNP/NSC-A. actinomycetemcomitans- Kills the adherent bacteria
- Anti-biofilm activity
- Control of peri-implantitis
[79]
Bismuth subsalicylate~4–22P. gingivalis
A. actinomycetemcomitans
- Antibacterial activity
- Application in dental materials
[80]
NACP: amorphous calcium phosphate NPs, MSN-PLGA: mesoporous silica nanoparticles (MSN) grafted with poly-L-glycolic acid (PGA), ZnO/Z: zinc oxide/zeolite nanocomposite, AgNPs-PL: silver nanoparticles poloxamer thermoreversible gel, CO: calcium oxide, CH: calcium hydroxide, MgO: nano-magnesium oxide, Ag/Zn-MCSNs: silver and zinc incorporated mesoporous calcium-silicate nanoparticles, PLLA@Ag: silver nanoparticle-loaded poly(L-lactide), AgCSP: chitosan/polyurethane (CSP) nanofibrous membrane incorporated silver nanoparticles, AgNP/NSC: silica-based composite coating containing starch-capped silver nanoparticles.
Table 2. A summary of nanoparticle applications in aPDT against oral bacterial infections.
Table 2. A summary of nanoparticle applications in aPDT against oral bacterial infections.
NanoparticlesSize (nm)PhotosensitizerLight SourceMicroorganismTherapeutic EffectsRef.
Nano-c-ICG810 nm light at an energy density of 4–24 J/cm2P. gingivalis- Bactericidal effects[93]
Chitosan~60Rose bengal260 nm light at an energy density of 4–60 J/cm2S. oralis,
P. intermedia,
A. naeslundii
- Increased affinity to bacterial cell membrane
- Anti-biofilm effects against multispecies bacterial biofilms
[94]
Fe3O4~122.4Chlorin e6630 nm light at an energy density of 100 J/cm2S. sanguinis
F. nucleatum
P. gingivalis
- Anti-biofilm activity

- Inhibits the occurrence and progression of periodontitis
[95]
Lys-Au- Rose bengalLEDS. mutans- Bacterial inactivation
- Anti-biofilm activity
[96]
Silver~18TBO630 nm light at an energy density of 100 J/cm2 S. mutans- Anti-biofilm activity
- Anti metabolic activity
[97]
HPMC-TBO630 nm light at an energy density of 32.4 J/cm2 S. aureus
A. actinomycetemcomitans
P. gingivalis
- Anti-biofilm activity[98]
UCNPs~25Chlorin e6980 nm light at an energy density of 750 J/cm2P. gingivalis
P. intermedia
F.nucleatum
- Anti-biofilm effects[83]
Liposomes~100–150tetra-m-hydroxyphenyl chlorin652 nm light at an energy density of 100 J/cm2E. faecalis- Antibacterial activity[99]
Nano-c: nanospheres coated with chitosan, ICG; indocyanine green, S. oralis: Streptococcus oralis, Lys-Au NCs: lysozyme-stabilized gold nanoclusters, LED: light-emitting diode, HPMC: chitosan hydrogels containing hydroxypropyl methylcellulose, UCNPs: upconversion nanoparticles (UCNPs) NaYF4:Yb,Er.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Afrasiabi, S.; Chiniforush, N.; Barikani, H.R.; Partoazar, A.; Goudarzi, R. Nanostructures as Targeted Therapeutics for Combating Oral Bacterial Diseases. Biomedicines 2021, 9, 1435. https://doi.org/10.3390/biomedicines9101435

AMA Style

Afrasiabi S, Chiniforush N, Barikani HR, Partoazar A, Goudarzi R. Nanostructures as Targeted Therapeutics for Combating Oral Bacterial Diseases. Biomedicines. 2021; 9(10):1435. https://doi.org/10.3390/biomedicines9101435

Chicago/Turabian Style

Afrasiabi, Shima, Nasim Chiniforush, Hamid Reza Barikani, Alireza Partoazar, and Ramin Goudarzi. 2021. "Nanostructures as Targeted Therapeutics for Combating Oral Bacterial Diseases" Biomedicines 9, no. 10: 1435. https://doi.org/10.3390/biomedicines9101435

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop